Layer-by-Layer Assembled Gold Nanoparticles for siRNA Delivery

Mar 30, 2009 - Layer-by-Layer Assembled Gold Nanoparticles for siRNA Delivery ..... Nanoparticles for Immunoassay Development: Toward Point of Care ...
57 downloads 0 Views 213KB Size
NANO LETTERS

Layer-by-Layer Assembled Gold Nanoparticles for siRNA Delivery

2009 Vol. 9, No. 5 2059-2064

Asmaa Elbakry,† Alaa Zaky,† Renate Liebl,† Reinhard Rachel,‡ Achim Goepferich,*,† and Miriam Breunig† Department of Pharmaceutical Technology and Centre for Electron Microscopy at the Institute for Anatomy, UniVersity of Regensburg, 93040 Regensburg, Germany Received February 5, 2009; Revised Manuscript Received March 20, 2009

ABSTRACT Although uptake into cells is highly complex and regulated, heterogeneous particle collectives are usually employed to deliver small interfering RNA (siRNA) to cells. Within these collectives, it is difficult to accurately identify the active species, and a decrease in efficacy is inherent to such preparations. Here, we demonstrate the manufacture of uniform nanoparticles with the deposition of siRNA on gold in a layer-by-layer approach, and we further report on the cellular delivery and siRNA activity as functions of surface properties.

The creation and design of nanosized carriers for the delivery of nucleic acids such as small interfering RNAs (siRNAs) have recently gained interest because of their possible clinical applications.1,2 The interaction with cell membranes is of central importance for these delivery systems, because this represents the key event in the regulation of the uptake process. The multiple portals of entry into mammalian cells strongly vary with regard to the nature and size of the cargo, as do their intracellular destination and fate.3 Consequently, the pathway of cellular entry will determine if the respective nucleic acid is unloaded at its site of action and if it is effective or not. Therefore, in order to deliver siRNA-loaded nanoparticles to specific intracellular destinations to elicit a distinctive biological effect, it is optimal to apply small, monodisperse nanoparticles with a defined ζ potential and surface chemistry. Although the field of siRNA therapeutics has made significant progress, the effective delivery of siRNA remains a challenge that must be addressed before clinical use of siRNA becomes a preferred technique.1,4,5 A plethora of materials that can form nanosized complexes with siRNA have previously been intensively investigated.6,7 Most studies have focused on cationic lipids and polymers, which form random self-assembled aggregates with nucleic acids that are often larger than 100 nm. These systems are generally heterogeneous and poorly defined particle collectives.8-11 For example, a commonly used system involves forming nanoparticles with poly(ethylene imine) (PEI) and nucleic acids. * To whom correspondence should be addressed. Phone: +49 941 943 4843. Fax: +49 941 943 4807. E-mail: achim.goepferich@chemie. uni-regensburg.de. Mailing Address: Universitaetsstrasse 31, 93040 Regensburg, Germany. † Department of Pharmaceutical Technology. ‡ Centre for Electron Microscopy at the Institute for Anatomy. 10.1021/nl9003865 CCC: $40.75 Published on Web 03/30/2009

 2009 American Chemical Society

The resulting collective contains different particle species that are in equilibrium with free polymer (about 86%).12 The heterogeneity of this system and the existence of different subpopulations in such random preparations may lead to a decrease in efficacy, and this also complicates the interpretation of experimental results tremendously since it is not evident which particular subspecies of the preparation is responsible for the overall biological effect. Furthermore, toxic effects that may be exhibited by only a fraction of the preparation may appear to be intrinsic properties of the whole collective. Because endocytosis is governed by highly sophisticated and well-regulated principles and because heterogeneous particle collectives are associated with all of the aforementioned disadvantages, it is clear that new strategies for the fabrication of efficient nanocarriers are essential to advance the field. While current approaches, which have focused on adjusting the size and surface properties of nanoparticles, are excellent models to help understand drug targeting principles and cellular uptake,13-17 they are not ideally suited to transport highly charged macromolecules such as siRNA. Still, gold nanoparticles (AuNPs) have preciously been used to deliver oligonucleotides or plasmid DNA18-20 and even siRNA21,22 into cells. Very recently, Mirkin and co-workers attached siRNA molecules to the surface of AuNPs via a thiol group.22 The polyvalent siRNA/nanoparticle conjugates showed a 6 times greater half-life and prolonged gene knockdown compared to free RNA duplexes. While these approaches demonstrate the general applicability of AuNPs as delivery vehicle, in some cases AuNPs seemed to aggregate after assembly with nucleic acids,18,21 the delivered nucleic acid showed low activity inside cells,20 or the efficacy of the nucleic acid relied on an additional transfection reagent.19

Scheme 1. Flowchart Illustrating the LbL Deposition Applied to AuNPsa

a After each coating step, AuNPs were purified by centrifugation as indicated and resuspended in unbuffered 10 mM NaCl.

We hypothesized that a layer-by-layer (LbL) strategy would allow us to use monodisperse AuNPs as a template for the manufacture of a carrier that remains monodisperse during assembly and delivers active siRNA into cells, thereby circumventing the limitations of previous systems. Although the LbL deposition of oppositely charged polyelectrolytes is an established method for the fabrication of thin films on flat solid surfaces and microparticles,23,24 the coating of nanoparticles presents a tremendous challenge. This is because many problems are associated with the wrapping of polyelectrolytes around nanoparticles with high curvature such as aggregation due to cross-linking of the particles by the polyelectrolyte chains and the separation of the unbound polyelectrolyte from the coated particles.25-27 Here, we present a first proof of concept that even small nanoparticles can be modified with therapeutically relevant siRNA molecules in a LbL approach, and we report on the experimental parameters of their fabrication as well as their cellular delivery. AuNPs as a core bear several advantages including straightforward synthesis, easy surface modification, availability in different size ranges with narrow size distribution, and finally high biocompatibility with cells or tissues.28-30 In this study, AuNPs were spherical, homogeneous, and free of aggregates. They had a size of 15.5 ( 1.2 nm as determined by transmission electron microscopy (TEM) (see Supporting Information, Figure S1). For the LbL coating of the nanoparticles, we envisioned a strategy that is illustrated in Scheme 1. First, 11mercaptoundecanoic acid (MUA) was to be deposited on the gold surface to facilitate the binding of the subsequent layers. Thereafter, the nanoparticles were to be consecutively added to oppositely charged polyelectrolyte solutions, first to PEI with a molecular weight of 25 kDa and then to doublestranded 21-mer siRNA. Finally, PEI was to complete the shell as a last layer. The major challenge involved identifying the appropriate parameters such that the coating and purification of AuNPs were possible. Although various polyelectrolytes have been applied for LbL deposition onto nanoparticles, siRNA was not expected to easily wrap around small AuNPs because it is a stiff, rodlike molecule.31 To obtain well-defined particles onto which the polyelectrolyte can be adsorbed with high yield and to avoid interparticle bridging and flocculation, it was important to choose the appropriate polyelectrolyte 2060

concentration and ionic strength.25-27,32 More specifically, smaller particles need a higher salt concentration to overcome the repulsive forces and to wrap the polymer around themselves. However, a high ionic strength may cause aggregation of gold nanoparticles.33 The optimal concentrations of PEI and siRNA during coating were determined to be 1.0 mg/ml and 2.0 µM, respectively (see Supporting Information, Figure S2). A final concentration of 10 mM NaCl proved to be suitable for maintaining a small hydrodynamic diameter and low polydispersity index of AuNPs during all coating and purification steps (see Supporting Information, Figure S3, exemplified for the resuspension of PEI-AuNPs after the second purification step). After each coating step, unbound polyelectrolyte was removed by centrifugation to avoid formation of nanoaggregates between free PEI and free siRNA, which would give rise to different, coexisting particle species. Two or three purification steps were necessary after coating with siRNA or PEI to ensure that only a minimal amount of polyelectrolyte was detectable in the supernatant (see Supporting Information, Table S1). The optimal parameters were applied to assemble the LbL particles with PEI and siRNA, and the successful polyelectrolyte deposition was monitored by dynamic light scattering (DLS), ζ-potential measurements, UV-vis absorbance spectra and TEM. Figure 1A details the change in the hydrodynamic diameter, including the polydispersity index as determined by DLS. The size of AuNPs increased during coating from 18.5 ( 0.2 nm (MUA-AuNPs) to 26.8 ( 0.3 nm (PEI/siRNA/PEI-AuNPs). Additionally, a reversal of the ζ-potential after completion of each layer signified the deposition of each polyelectrolyte (Figure 1B). After purification, the ζ-potential slightly decreased due to removal of unbound polyelectrolyte. The red shift of the surface plasmon band of gold nanoparticles was used to distinguish between polymer adsorption onto the AuNP surface and interparticulate bridging and aggregation.25-27,32 The UV-vis spectra in Figure 1C revealed that the AuNPs predominantly remained disaggregated after coating and that only low interparticulate bridging could have occurred, as the maximum of the plasmon peak shifted between 1 and 2 nm with consecutive layer deposition (Figure 1D). This was confirmed by statistical evaluation of electron micrographs (Figure 1E) that were made after each layer during LBL assembly (500 AuNPs were measured for each type of AuNPs; for micrographs, please refer to Supporting Information, Figure S1). The majority of AuNPs remained as single particles during the LbL assembly. Although an increasing amount of AuNP doublets (2 Au cores per aggregate) appeared with consecutive layer build-up, it never surpassed a fraction larger than 7%. Also worth noting is the number of siRNA molecules per AuNP, which was calculated to be around 780 siRNA molecules per siRNA/PEI-AuNP and PEI/siRNA/PEI-AuNP (for calculation refer to Supporting Information). This number is in a similar range as the one for AuNPs that were functionalized with 15-mer oligonucleotides via a thiol linker.34 Overall, these results demonstrated that by selecting the experimental parameters accordingly during LbL Nano Lett., Vol. 9, No. 5, 2009

Figure 1. Characterization of AuNPs during LbL build-up by different methods. (A) The effect of the subsequent coating steps on the hydrodynamic diameter (white columns) and the polydispersity index (rhombs) of AuNPs before (crude) and after purification (purified) are shown as well as (B) the corresponding ζ-potential. The values represent the mean plus/minus standard deviation of three subruns of sample. One representative experiment of five independent ones is shown. (C) UV-vis spectra of AuNPs from top to bottom: MUAAuNPs, PEI-AuNPs, siRNA/PEI-AuNPs, PEI/siRNA/PEI-AuNPs. (D) The corresponding maximum position of the plasmon resonance peak. (E) Quantitative evaluation of TEM images (see Supporting Information, Figure S1). 500 AuNPs were counted and the fraction of n Au-cores per aggregate was determined. The number of Au cores was n ) 1 (white columns) or n ) 2 (black columns).

assembly, most AuNPs remained single and maintained their small and uniform size. Random, self-assembled aggregates of PEI and nucleic acids may suffer from severe aggregation when subjected to the high ion strength of biological fluids.8,35 As a result of aggregation, the initial size of these complexes may increase to several micrometers.35 This feature would make the fabrication of LbL-coated nanoparticles tedious and would render their application in cell culture pointless. Hence, AuNPs were exposed to cell culture medium containing fetal bovine serum, and the hydrodynamic diameter and the ζ-potential were monitored (Figure 2A,B). Irrespective of the type of the outer layer (PEI or siRNA), the hydrodynamic diameter of AuNPs increased only by a factor of 2.1 to 2.5. This increase in diameter is also reflected in the corresponding UV-vis spectra shown in Figure 2C (maximum of the plasmon peak was as follows: PEI-AuNPs, 530 nm, siRNA/PEI-AuNPs, 532 nm; PEI/siRNA/PEI-AuNPs, 536 nm). The extent of growth in diameter can most likely be attributed to the adsorption of proteins on the surface, as already shown for unmodified and other oligonucleotide modified AuNPs.16,36 Interestingly, the ζ-potential of all particle types was negative after incubation with serumcontaining culture medium (Figure 2B). More specifically, Nano Lett., Vol. 9, No. 5, 2009

Figure 2. Characterization of AuNPs in different media. AuNPs were diluted in Millipore water (9) or serum-containing culture medium (0) and (A) the particle size as well as (B) the ζ-potential were measured. The values represent the mean plus/minus standard deviation of three subruns of one sample. One representative experiment of three independent ones is shown. The UV-vis spectra of AuNPs after dilution in serum-containing (C) or serumfree culture medium (D). The spectra from top to bottom are as follows: PEI-AuNPs, siRNA/PEI-AuNPs, PEI/siRNA/PEI-AuNPs.

the negative ζ-potential of siRNA/PEI-AuNPs became more positive, while PEI- and PEI/siRNA/PEI-AuNPs showed a 2061

Figure 3. The cellular uptake of AuNPs. The number of AuNPs per cell as determined by ICP-OES and cell counting (for details of calculation refer to Supporting Information) after 6 h of incubation in serum-containing culture medium as a function of surface modification. The initial concentration of AuNPs in the culture medium was 0.32 and 0.30 nM for siRNA/PEI- and PEI/ siRNA/PEI-AuNPs, respectively. For each measurement cells from 3 culture flasks (75 cm2 each) were pooled to obtain values within the calibration range, bars are averages of two measurements of one representative experiment of two independent ones.

charge reversal. After incubation in serum-free culture medium, the particle size increased by 7 to 10-fold (data not shown), and the UV-vis spectra in Figure 2D show that the individuality of AuNPs was lost (maximum of the plasmon peak was as follows: PEI-AuNPs: 575 nm, siRNA/ PEI-AuNPs: 580 nm, PEI/siRNA/PEI-AuNPs: 582 nm), which corroborates the hypothesis that serum proteins are necessary for stabilization of the nanoparticles. The positive surface charge of self-assembled complexes of PEI and nucleic acids guarantees the cellular entry of negatively charged nucleic acids that are usually not incorporated by cells.35 Therefore, it was not evident that LbLassembled nanoparticles with an overall negative surface charge, even with PEI as a surface layer, were taken up by cells. To obtain quantitative data for the number of AuNPs per cell, CHO-K1 cells were incubated for 6 h with siRNA/ PEI- or PEI/siRNA/PEI-AuNPs in serum-containing culture medium, and subsequently the Au3+ content of the cell digest was determined by inductively coupled plasma spectroscopy (ICP-OES). Both types of particles were successfully taken up by CHO-K1 cells and the average amount of AuNPs per cell strongly depended on the type of the top layer. CHOK1 cells internalized more siRNA/PEI-AuNPs (estimated number, 2.0 × 105 per cell) in relation to PEI/siRNA/PEIAuNPs (estimated number, 5.6 × 104 per cell) (Figure 3). This difference correlated well with the electron micrographs of sections of CHO-K1 cells (see Supporting Information, Figures S4 and S5). However, it must be noted that no distinction between AuNPs that entered the cells and those stuck on the outside on the cell membrane was made in this method. Transmission electron micrographs of CHO-K1 cells (see Supporting Information, Figures S4 and S5) illustrate that this consideration must be taken into account for PEI/ siRNA/PEI-AuNPs, but not for siRNA/PEI-AuNPs. Therefore, the ICP-OES and the transmission electron micrographs together allow for the conclusion that the number of siRNA/ PEI-AuNPs per cell is significantly higher as compared to PEI/siRNA/PEI-AuNPs. In comparison to citrate-stabilized 2062

Figure 4. The cellular uptake of (A-C) siRNA/PEI-AuNPs and (D-F) PEI/siRNA/PEI-AuNPs into CHO-K1 cells as evaluated by TEM. The bar indicates 0.13 µm in D and 0.20 µm in all other images.

AuNPs, the amount of siRNA/PEI- and PEI/siRNA/PEIAuNPs per cell was much higher,16 while the number of siRNA/PEI-AuNPs per cell was similar to other DNAmodified AuNPs.36 The significant difference in cellular uptake of AuNPs that are of nearly similar size but possess different outer layers suggests that surface properties may strongly affect interactions with cells. Their homogeneous size distribution renders the AuNPs highly favorable for this investigation, because it has been shown that the size of AuNPs strongly influences the number of AuNPs that are taken up per cell.16 Hence, we can conclude that the differences in the cellular uptake of siRNA/PEI- and PEI/siRNA/PEI-AuNPs are due to differences in surface properties and not in size (due to a broad size distribution). Additionally, we show electron micrographs of sectioned CHO-K1 cells that help to elucidate the intracellular fate of the AuNPs. The micrographs in Figure 4 illustrate that the AuNPs were predominantly trapped within endocytotic vesicles after 6 h of incubation (micrographs at magnification are included into Supporting Information, Figure S4 and S5), and no particles were detected in the nucleus. Figures 4A-C show typical images after incubation with siRNA/PEI-AuNPs. Each endocytotic vesicle contained several nanoparticles. Within one vesicle, some particles were still single or have formed a doublet or a triplet (3 Au cores per aggregate), but most have aggregated to larger clusters. In contrast, after incubation with PEI/ siRNA/PEI-AuNPs (Figure 4D-F), less vesicles per cell containing AuNPs were counted, and the number of AuNPs per vesicle was also lower as compared to siRNA/PEI-coated particles. It was remarkable that inside of the endocytotic vesicles, the particles remained either single or formed at most doublets. Some PEI/siRNA/PEI-AuNPs, but not siRNA/ PEI-AuNPs, were also detected in caveolae-like structures (Figure 4D). It will be of great interest to investigate the intracellular fate of PEI/siRNA/PEI-AuNPs and siRNA/PEIAuNPs that are produced in different size ranges in future experiments. Having demonstrated that the AuNPs are uniform in size and surface charge and homogeneous in distribution, we Nano Lett., Vol. 9, No. 5, 2009

Figure 5. (A) Gene silencing of EGFP in CHO-K1 cells stably expressing EGFP and (B) relative cell viability after addition of PEI/siRNA/PEI-AuNPs at various initial concentrations. For LbL assembly of nanoparticles either siRNA against EGFP (9) was used or a nontargeted siRNA control (0). The values represent the mean plus/minus standard deviation (n ) 3). One representative experiment of three independent ones is shown.

wanted to determine their intracellular fate as siRNA carriers. In particular, it was of great importance to provide evidence of activity of the siRNA inside the cells. This capability was tested in CHO-K1 cells stably expressing enhanced green fluorescent protein (CHO-K1/EGFP). To test the specificity of the knock-down, two different types of siRNA were applied for LbL-nanoparticle formation: siRNA against EGFP and a nontargeted siRNA sequence as control. A dosedependent knock-down was measured for PEI/siRNA/PEIAuNPs, and the cellular EGFP production was reduced to about 28% (Figure 5A). Control formulations with nontargeted siRNA revealed that gene silencing was highly specific. The cell viability of CHO-K1/EGFP cells was not seriously influenced by the addition of AuNPs (Figure 5B), as it only slightly decreased to 93.4 ( 1.1% and 95.0 ( 0.8%, respectively, at the highest AuNP concentration. When siRNA was used as the top layer, no silencing efficacy was detected (see Supporting Information, Figure S6). Possible reasons may include degradation of unprotected siRNA on the surface of the nanoparticles, inadequate endosomal escape of nanoparticles due to a lower amount of the transfection reagent per particle or even the agglomeration of the AuNPs during cellular uptake. We also compared the parameters of siRNA delivery using PEI/siRNA/PEI-AuNPs with the ones using random-assembled siRNA/PEI-agglomerates. For the same amount of cells, the latter process usually requires 100 nM siRNA in combination with 3.8 to 5.1 µg PEI to achieve a similar knock-down in gene expression.37 In this study, both the siRNA and PEI concentration were higher (siRNA, 1.2-2.9 fold; PEI, 12-16 fold). The higher siRNA concentration may most likely be necessary because LbL-coated AuNPs are very Nano Lett., Vol. 9, No. 5, 2009

stable and, hence, the release of siRNA inside cells may occur only to a low extent. Therefore, in future experiments the fabrication of LbL nanoparticles with polymers that are biodegradable inside cells will most likely allow for improved release of siRNA and, subsequently, a lower AuNP concentration will be sufficient to achieve the same effect. It was remarkable that PEI/siRNA/PEI-AuNPs showed no serious toxicity in cell culture despite a PEI concentration that typically induces devastating cellular toxicity. One possible reason may be the removal of free polymer, as this is usually associated with the high toxicity of PEI.38-40 In summary, we were the first to successfully show the LbL assembly of the oppositely charged polyelectrolytes siRNA and PEI on AuNPs. This technique offers a unique opportunity to fabricate well-defined and homogenously distributed nanocarriers for siRNA delivery. In future experiments, these LbL-coated nanoparticles will provide an excellent tool to study how the size and the surface properties influence the portal of entry into cells and direct distinct particles to the correct site of activity. The obtained information will help to detect existing limitations and to design new materials for siRNA delivery. Acknowledgment. The authors thank Cornelia Rose, Angelika Berie´, and Dr. Harald Huber for ICP analysis, Angelika Ku¨hn for support with EM preparation, and Siddharth Pathi from Cornell University for proofreading the manuscript. Supporting Information Available: Detailed description of methods; TEM images of AuNPs; hydrodynamic diameter of AuNPs at various PEI and siRNA concentrations and at different ionic strengths; TEM pictures of CHO-K1 cells; gene silencing of siRNA/PEI-AuNPs; concentration of PEI and siRNA in the supernatant after purification. This material is available free of charge via the Internet at http:// pubs.acs.org. References (1) Aagaard, L.; Rossi, J. J. AdV. Drug DeliVery ReV. 2007, 59, 75–86. (2) Dykxhoorn, D. M.; Palliser, D.; Lieberman, J. Gene Ther. 2006, 13, 541–552. (3) Conner, S. D.; Schmid, S. L. Nature (London) 2003, 422, 37–44. (4) Whitehead, K. A.; Langer, R.; Anderson, D. G. Nat. ReV. Drug DiscoV. 2009, 8, 129–138. (5) Juliano, R.; Alam, M.; Dixit, V.; Kang, H. Nucleic Acids Res. 2008, 36, 4158–4171. (6) Zhang, S.; Zhao, B.; Jiang, H.; Wang, B.; Ma, B. J. Controlled Release 2007, 123, 1–10. (7) de Fougerolles, A.; Vornlocher, H. P.; Maraganore, J.; Lieberman, J. Nat. ReV. Drug DiscoVery 2007, 6, 443–453. (8) Neu, M.; Fischer, D.; Kissel, T. J. Gene Med. 2005, 7, 992–1009. (9) Pozharski, E. V.; MacDonald, R. C. Mol. Pharm. 2007, 4, 962–974. (10) Chesnoy, S.; Huang, L. Annu. ReV. Biophys. Biomol. Struct. 2000, 29, 27–47. (11) Elouahabi, A.; Ruysschaert, J. M. Mol. Ther. 2005, 11, 336–347. (12) Clamme, J. P.; Azoulay, J.; Mely, Y. Biophys. J. 2003, 84, 1960– 1968. (13) Hild, W. A.; Breunig, M.; Goepferich, A. Eur. J. Pharm. Biopharm. 2008, 68, 153–168. (14) Cambi, A.; Lidke, D. S.; Arndt-Jovin, D. J.; Figdor, C. G.; Jovin, T. M. Nano Lett. 2007, 7, 970–977. (15) Javier, D. J.; Nitin, N.; Levy, M.; Ellington, A.; Richards-Kortum, R. Bioconjug. Chem. 2008, 19, 1309–1312. (16) Chithrani, B. D.; Ghazani, A. A.; Chan, W. C. Nano Lett. 2006, 6, 662–668. 2063

(17) Bagalkot, V.; Zhang, L.; Levy-Nissenbaum, E.; Jon, S.; Kantoff, P. W.; Langer, R.; Farokhzad, O. C. Nano Lett. 2007, 7, 3065–3070. (18) Thomas, M.; Klibanov, A. M. Proc. Natl. Acad. Sci. U.S.A. 2003, 100, 9138–9143. (19) Liu, Y.; Franzen, S. Bioconjug. Chem. 2008, 19, 1009–1016. (20) Rosi, N. L.; Giljohann, D. A.; Thaxton, C. S.; Lytton-Jean, A. K. R.; Han, M. S.; Mirkin, C. A. Science 2006, 312, 1027–1030. (21) Lee, S. H.; Bae, K. H.; Kim, S. H.; Lee, K. R.; Park, T. G. Int. J. Pharm. 2008, 364, 94–101. (22) Giljohann, D. A.; Seferos, D. S.; Prigodich, A. E.; Patel, P. C.; Mirkin, C. A. J. Am. Chem. Soc. 2009, 131, 2072–2073. (23) Decher, G.; Schlenoff, J. Multilayer thin films; Wiley-VCH: Weinheim, 2002. (24) Tang, Z.; Wang, Y.; Podsiadlo, P.; Kotov, N. A. AdV. Mater. 2006, 18, 3203–3224. (25) Gittins, D. I.; Caruso, F. J. Phys. Chem. B 2001, 105, 6846–6852. (26) Mayya, K. S.; Schoeler, B.; Caruso, F. AdV. Funct. Mater. 2003, 13, 183–188. (27) Schneider, G.; Decher, G. Langmuir 2008, 24, 1778–1789. (28) Daniel, M. C.; Astruc, D. Chem. ReV. 2004, 104, 293–346. (29) Shukla, R.; Bansal, V.; Chaudhary, M.; Basu, A.; Bhonde, R. R.; Sastry, M. Langmuir 2005, 21, 10644–10654.

2064

(30) Ghosh, P.; Han, G.; De, M.; Kim, C. K.; Rotello, V. M. AdV. Drug DeliVery ReV. 2008, 60, 1307–1315. (31) Gary, D. J.; Puri, N.; Won, Y. Y. J. Controlled Release 2007, 121, 64–73. (32) Schneider, G.; Decher, G. Nano Lett. 2004, 4, 1833–1839. (33) Hayat, M. A. Colloidal gold: principles, methods and applications; Academic Press: San Diego, 1989. (34) Hurst, S. J.; Lytton-Jean, A. K. R.; Mirkin, C. A. Anal. Chem. 2006, 78, 8313–8318. (35) Lungwitz, U.; Breunig, M.; Blunk, T.; Goepferich, A. Eur. J. Pharm. Biopharm. 2005, 60, 247–266. (36) Giljohann, D. A.; Seferos, D. S.; Patel, P. C.; Millstone, J. E.; Rosi, N. L.; Mirkin, C. A. Nano Lett. 2007, 7, 3818–3821. (37) Breunig, M.; Hozsa, C.; Lungwitz, U.; Watanabe, K.; Umeda, I.; Kato, H.; Goepferich, A. J. Controlled Release 2008, 130, 57–63. (38) Boeckle, S.; von Gersdorff, K.; van der, P. S.; Culmsee, C.; Wagner, E.; Ogris, M. J. Gene Med. 2004, 6, 1102–1111. (39) Moghimi, S. M.; Symonds, P.; Murray, J. C.; Hunter, A. C.; Debska, G.; Szewczyk, A. Mol. Ther. 2005, 11, 990–995. (40) Chollet, P.; Favrot, M. C.; Hurbin, A.; Coll, J. L. J. Gene Med. 2002, 4, 84–91.

NL9003865

Nano Lett., Vol. 9, No. 5, 2009